101
|
Folegatti PM, Jenkin D, Morris S, Gilbert S, Kim D, Robertson JS, Smith ER, Martin E, Gurwith M, Chen RT, For the Benefit-Risk Assessment of VAccines by TechnolOgy Working Group BRAVATO, ex-V3SWG). Vaccines based on the replication-deficient simian adenoviral vector ChAdOx1: Standardized template with key considerations for a risk/benefit assessment. Vaccine 2022; 40:5248-5262. [PMID: 35715352 PMCID: PMC9194875 DOI: 10.1016/j.vaccine.2022.06.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/10/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023]
Abstract
Replication-deficient adenoviral vectors have been under investigation as a platform technology for vaccine development for several years and have recently been successfully deployed as an effective COVID-19 counter measure. A replication-deficient adenoviral vector based on the simian adenovirus type Y25 and named ChAdOx1 has been evaluated in several clinical trials since 2012. The Brighton Collaboration Benefit-Risk Assessment of VAccines by TechnolOgy (BRAVATO) was formed to evaluate the safety and other key features of new platform technology vaccines. This manuscript reviews key features of the ChAdOx1-vectored vaccines. The simian adenovirus Y25 was chosen as a strategy to circumvent pre-existing immunity to common human adenovirus serotypes which could impair immune responses induced by adenoviral vectored vaccines. Deletion of the E1 gene renders the ChAdOx1 vector replication incompetent and further genetic engineering of the E3 and E4 genes allows for increased insertional capability and optimizes vaccine manufacturing processes. ChAdOx1 vectored vaccines can be manufactured in E1 complementing cell lines at scale and are thermostable. The first ChAdOx1 vectored vaccines approved for human use, against SARS-CoV-2, received emergency use authorization in the UK on 30th December 2020, and is now approved in more than 180 countries. Safety data were compiled from phase I-III clinical trials of ChAdOx1 vectored vaccines expressing different antigens (influenza, tuberculosis, malaria, meningococcal B, prostate cancer, MERS-CoV, Chikungunya, Zika and SARS-CoV-2), conducted by the University of Oxford, as well as post marketing surveillance data for the COVID-19 Oxford-AstraZeneca vaccine. Overall, ChAdOx1 vectored vaccines have been well tolerated. Very rarely, thrombosis with thrombocytopenia syndrome (TTS), capillary leak syndrome (CLS), immune thrombocytopenia (ITP), and Guillain-Barre syndrome (GBS) have been reported following mass administration of the COVID-19 Oxford-AstraZeneca vaccine. The benefits of this COVID-19 vaccination have outweighed the risks of serious adverse events in most settings, especially with mitigation of risks when possible. Extensive immunogenicity clinical evaluation of ChAdOx1 vectored vaccines reveal strong, durable humoral and cellular immune responses to date; studies to refine the COVID-19 protection (e.g., via homologous/heterologous booster, fractional dose) are also underway. New prophylactic and therapeutic vaccines based on the ChAdOx1 vector are currently undergoing pre-clinical and clinical assessment, including vaccines against viral hemorrhagic fevers, Nipah virus, HIV, Hepatitis B, amongst others.
Collapse
Affiliation(s)
| | | | | | | | - Denny Kim
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - James S. Robertson
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Emily R. Smith
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA,Corresponding author
| | - Emalee Martin
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Marc Gurwith
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Robert T. Chen
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | | |
Collapse
|
102
|
Bigay J, Le Grand R, Martinon F, Maisonnasse P. Vaccine-associated enhanced disease in humans and animal models: Lessons and challenges for vaccine development. Front Microbiol 2022; 13:932408. [PMID: 36033843 PMCID: PMC9399815 DOI: 10.3389/fmicb.2022.932408] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
The fight against infectious diseases calls for the development of safe and effective vaccines that generate long-lasting protective immunity. In a few situations, vaccine-mediated immune responses may have led to exacerbated pathology upon subsequent infection with the pathogen targeted by the vaccine. Such vaccine-associated enhanced disease (VAED) has been reported, or at least suspected, in animal models, and in a few instances in humans, for vaccine candidates against the respiratory syncytial virus (RSV), measles virus (MV), dengue virus (DENV), HIV-1, simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), and the Middle East respiratory syndrome coronavirus (MERS-CoV). Although alleviated by clinical and epidemiological evidence, a number of concerns were also initially raised concerning the short- and long-term safety of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is causing the ongoing COVID-19 pandemic. Although the mechanisms leading to this phenomenon are not yet completely understood, the individual and/or collective role of antibody-dependent enhancement (ADE), complement-dependent enhancement, and cell-dependent enhancement have been highlighted. Here, we review mechanisms that may be associated with the risk of VAED, which are important to take into consideration, both in the assessment of vaccine safety and in finding ways to define models and immunization strategies that can alleviate such concerns.
Collapse
Affiliation(s)
| | | | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud-INSERM U1184, CEA, Fontenay-Aux-Roses, France
| | | |
Collapse
|
103
|
LAMP-1 Chimeric to HIV-1 p55Gag in the Immunization of Neonate Mice Induces an Early Germinal Center Formation and AID Expression. Vaccines (Basel) 2022; 10:vaccines10081246. [PMID: 36016134 PMCID: PMC9414238 DOI: 10.3390/vaccines10081246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Neonates have a limited adaptive response of plasma cells, germinal center (GC) B cells, and T follicular helper cells (TFH). As neonatal vaccination can be an important tool for AIDS prevention, these limitations need to be overcome. Chimeric DNA vaccine encoding p55Gag HIV-1 protein conjugated with lysosomal-associated membrane protein 1 (LAMP-1) has been described as immunogenic in the neonate period. Herein, we investigated the immunologic mechanisms involved in neonatal immunization with a LAMP-1/p55Gag (LAMP/Gag) DNA vaccine in a C57BL/6 mouse background. Neonatal LAMP/Gag vaccination induced strong Gag-specific T-cell response until adulthood and elevated levels of anti-Gag IgG antibodies. We also demonstrated for the first time that the immunogenicity of the neonatal period with LAMP/Gag is due to the induction of high-affinity anti-p24 IgG antibodies and long-term plasma cells. Together with that, there is the generation of early TFH cells and the formation of GC sites with the upregulation of activation-induced cytidine deaminase (AID) enzyme mRNA and protein expression in draining lymph nodes after neonatal LAMP/Gag vaccination. These findings underscore that the LAMP-1 strategy in the chimeric vaccine could be useful to enhance antibody production even in the face of neonatal immaturity, and they contribute to the development of new vaccine approaches for other emerging pathogens at an early stage of life.
Collapse
|
104
|
McCann N, O'Connor D, Lambe T, Pollard AJ. Viral vector vaccines. Curr Opin Immunol 2022; 77:102210. [PMID: 35643023 PMCID: PMC9612401 DOI: 10.1016/j.coi.2022.102210] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 01/06/2023]
Abstract
Over the past two years, the SARS-CoV-2 pandemic has highlighted the impact that emerging pathogens can have on global health. The development of new and effective vaccine technologies is vital in the fight against such threats. Viral vectors are a relatively new vaccine platform that relies on recombinant viruses to deliver selected immunogens into the host. In response to the SARS-CoV-2 pandemic, the development and subsequent rollout of adenoviral vector vaccines has shown the utility, impact, scalability and efficacy of this platform. Shown to elicit strong cellular and humoral immune responses in diverse populations, these vaccine vectors will be an important approach against infectious diseases in the future.
Collapse
Affiliation(s)
- Naina McCann
- Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Headington, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Daniel O'Connor
- Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Headington, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Headington, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Headington, Oxford OX3 7LE, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
105
|
Cheng H, Peng Z, Si S, Alifu X, Zhou H, Chi P, Zhuang Y, Mo M, Yu Y. Neutralization Activity against SARS-CoV-2 Variants after Booster Vaccination in Populations without COVID-19: A Meta-Analysis. Vaccines (Basel) 2022; 10:1101. [PMID: 35891263 PMCID: PMC9322873 DOI: 10.3390/vaccines10071101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 02/04/2023] Open
Abstract
A number of SARS-CoV-2 variants that have evolved to have significant immune escape have emerged worldwide since the COVID-19 outbreak. The efficacy of prime vaccination is waning with the evolution of SARS-CoV-2, and the necessity of booster doses is more and more prominent. Therefore, this study aimed to compare the neutralization activity against the wild type and variants (Beta, Delta, and Omicron) in different prime-boost vaccination regimens. Electronic databases including PubMed, the Cochrane Library, Embase, medRxiv, Wanfang and CNKI were used to retrieve original studies. A total of 16 studies, 9 prime-boost vaccination regimes, and 3134 subjects were included in the meta-analysis and random effect models were used to estimate pooled neutralization titers. The neutralization activity against SARS-CoV-2 showed a significant decline with the evolution of the virus, especially in the populations primed with inactivated vaccines. For homologous immunization, only the populations boosted with mRNA vaccines consistently had a significant rise in neutralization titers (Beta: MD = 0.97; Delta: MD = 1.33; Omicron: MD = 0.74). While the heterologous immunization was more effective, the increment of neutralization titers against wild type, Beta, Delta and Omicron was 1.65 (95% CI: 1.32-1.96), 1.03 (95% CI: 0.53-1.54), 1.46 (95% CI: 1.07-1.85) and 1.15 (95% CI: 0.68-1.61), respectively. With the evolution of SARS-CoV-2, the effectiveness of prime immunization is waning. Although the administration of the booster dose could ameliorate the neutralization titers, homologous immunization regimens were gradually losing their effectiveness. Therefore, a heterologous booster dose is required, especially in populations primed with inactivated vaccines.
Collapse
Affiliation(s)
- Haoyue Cheng
- Department of Public Health and Department of Anesthesiology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health and Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhicheng Peng
- Department of Public Health and Department of Anesthesiology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health and Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shuting Si
- Department of Public Health and Department of Anesthesiology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health and Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xialidan Alifu
- Department of Public Health and Department of Anesthesiology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health and Medicine, Zhejiang University, Hangzhou 310058, China
| | - Haibo Zhou
- Department of Public Health and Department of Anesthesiology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health and Medicine, Zhejiang University, Hangzhou 310058, China
| | - Peihan Chi
- Department of Public Health and Department of Anesthesiology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health and Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yan Zhuang
- Department of Public Health and Department of Anesthesiology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health and Medicine, Zhejiang University, Hangzhou 310058, China
| | - Minjia Mo
- Department of Public Health and Department of Anesthesiology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health and Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yunxian Yu
- Department of Public Health and Department of Anesthesiology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; (H.C.); (Z.P.); (S.S.); (X.A.); (H.Z.); (P.C.); (Y.Z.); (M.M.)
- Department of Epidemiology & Health Statistics, School of Public Health and Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
106
|
Dangi T, Sanchez S, Lew MH, Visvabharathy L, Richner J, Koralnik IJ, Penaloza-MacMaster P. Pre-existing immunity modulates responses to mRNA boosters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.06.27.497248. [PMID: 35794898 PMCID: PMC9258286 DOI: 10.1101/2022.06.27.497248] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
mRNA vaccines have shown high efficacy in preventing severe COVID-19, but breakthrough infections, emerging variants and waning antibody levels have warranted the use of boosters. Although mRNA boosters have been widely implemented, the extent to which pre-existing immunity influences the efficacy of boosters remains unclear. In a cohort of individuals primed with the mRNA-1273 or BNT162b2 vaccines, we observed that lower antibody levels before boost were associated with higher fold-increase in antibody levels after boost, suggesting that pre-existing antibody modulates the boosting capacity of mRNA vaccines. Mechanistic studies in mice show that pre-existing antibodies significantly limit antigen expression and priming of B cell responses after mRNA vaccination. Furthermore, we demonstrate that the relative superiority of an updated Omicron vaccine over the original vaccine is critically dependent on the serostatus of the host. These data demonstrate that pre-existing immunity dictates responses to mRNA vaccination, elucidating specific circumstances when updated SARS-CoV-2 vaccines confer superior protection to original vaccines.
Collapse
Affiliation(s)
- Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Min Han Lew
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lavanya Visvabharathy
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Justin Richner
- Department of Microbiology & Immunology, University of Illinois Chicago College of Medicine, Chicago, IL 60612
| | - Igor J. Koralnik
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
107
|
Gene Therapy for Mitochondrial Diseases: Current Status and Future Perspective. Pharmaceutics 2022; 14:pharmaceutics14061287. [PMID: 35745859 PMCID: PMC9231068 DOI: 10.3390/pharmaceutics14061287] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial diseases (MDs) are a group of severe genetic disorders caused by mutations in the nuclear or mitochondrial genome encoding proteins involved in the oxidative phosphorylation (OXPHOS) system. MDs have a wide range of symptoms, ranging from organ-specific to multisystemic dysfunctions, with different clinical outcomes. The lack of natural history information, the limits of currently available preclinical models, and the wide range of phenotypic presentations seen in MD patients have all hampered the development of effective therapies. The growing number of pre-clinical and clinical trials over the last decade has shown that gene therapy is a viable precision medicine option for treating MD. However, several obstacles must be overcome, including vector design, targeted tissue tropism and efficient delivery, transgene expression, and immunotoxicity. This manuscript offers a comprehensive overview of the state of the art of gene therapy in MD, addressing the main challenges, the most feasible solutions, and the future perspectives of the field.
Collapse
|
108
|
Adenovirus DNA Polymerase Loses Fidelity on a Stretch of Eleven Homocytidines during Pre-GMP Vaccine Preparation. Vaccines (Basel) 2022; 10:vaccines10060960. [PMID: 35746566 PMCID: PMC9227658 DOI: 10.3390/vaccines10060960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/09/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022] Open
Abstract
In this study, we invented and construct novel candidate HIV-1 vaccines. Through genetic and protein engineering, we unknowingly constructed an HIV-1-derived transgene with a homopolymeric run of 11 cytidines, which was inserted into an adenovirus vaccine vector. Here, we describe the virus rescue, three rounds of clonal purification and preparation of good manufacturing practise (GMP) starting material assessed for genetic stability in five additional virus passages. Throughout these steps, quality control assays indicated the presence of the transgene in the virus genome, expression of the correct transgene product and immunogenicity in mice. However, DNA sequencing of the transgene revealed additional cytidines inserted into the original 11-cytidine region, and the GMP manufacture had to be aborted. Subsequent analyses indicated that as little as 1/25th of the virus dose used for confirmation of protein expression (106 cells at a multiplicity of infection of 10) and murine immunogenicity (108 infectious units per animal) met the quality acceptance criteria. Similar frameshifts in the expressed proteins were reproduced in a one-reaction in vitro transcription/translation employing phage T7 polymerase and E. coli ribosomes. Thus, the most likely mechanism for addition of extra cytidines into the ChAdOx1.tHIVconsv6 genome is that the adenovirus DNA polymerase lost its fidelity on a stretch of 11 cytidines, which informs future adenovirus vaccine designs.
Collapse
|
109
|
Richert L, Lelièvre JD, Lacabaratz C, Hardel L, Hocini H, Wiedemann A, Lucht F, Poizot-Martin I, Bauduin C, Diallo A, Rieux V, Rouch E, Surenaud M, Lefebvre C, Foucat E, Tisserand P, Guillaumat L, Durand M, Hejblum B, Launay O, Thiébaut R, Lévy Y. T Cell Immunogenicity, Gene Expression Profile, and Safety of Four Heterologous Prime-Boost Combinations of HIV Vaccine Candidates in Healthy Volunteers: Results of the Randomized Multi-Arm Phase I/II ANRS VRI01 Trial. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2663-2674. [PMID: 35613727 DOI: 10.4049/jimmunol.2101076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/03/2022] [Indexed: 06/15/2023]
Abstract
Heterologous prime-boost strategies are of interest for HIV vaccine development. The order of prime-boost components could be important for the induction of T cell responses. In this phase I/II multi-arm trial, three vaccine candidates were used as prime or boost: modified vaccinia Ankara (MVA) HIV-B (coding for Gag, Pol, Nef); HIV LIPO-5 (five lipopeptides from Gag, Pol, Nef); DNA GTU-MultiHIV B (coding for Rev, Nef, Tat, Gag, Env gp160 clade B). Healthy human volunteers (n = 92) were randomized to four groups: 1) MVA at weeks 0/8 + LIPO-5 at weeks 20/28 (M/L); 2) LIPO-5 at weeks 0/8 + MVA at weeks 20/28 (L/M); 3) DNA at weeks 0/4/12 + LIPO-5 at weeks 20/28 (G/L); 4) DNA at weeks 0/4/12 + MVA at weeks 20/28 (G/M). The frequency of IFN-γ-ELISPOT responders at week 30 was 33, 43, 0, and 74%, respectively. Only MVA-receiving groups were further analyzed (n = 62). Frequency of HIV-specific cytokine-positive (IFN-γ, IL-2, or TNF-α) CD4+ T cells increased significantly from week 0 to week 30 (median change of 0.06, 0.11, and 0.10% for M/L, L/M, and G/M, respectively), mainly after MVA vaccinations, and was sustained until week 52. HIV-specific CD8+ T cell responses increased significantly at week 30 in M/L and G/M (median change of 0.02 and 0.05%). Significant whole-blood gene expression changes were observed 2 wk after the first MVA injection, regardless of its use as prime or boost. An MVA gene signature was identified, including 86 genes mainly related to cell cycle pathways. Three prime-boost strategies led to CD4+ and CD8+ T cell responses and to a whole-blood gene expression signature primarily due to their MVA HIV-B component.
Collapse
Affiliation(s)
- Laura Richert
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR1219, Bordeaux, France
- Inria SISTM Team, Talence, France
- CHU de Bordeaux, Service d'Information Médicale, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Jean-Daniel Lelièvre
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
- Groupe Henri-Mondor Albert-Chenevier, AP-HP, Créteil, France
| | - Christine Lacabaratz
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Lucile Hardel
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR1219, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Hakim Hocini
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Aurélie Wiedemann
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Frédéric Lucht
- CHU de Saint Etienne, Saint-Priest-en-Jarez, France
- Université Jean Monnet and Université de Lyon, Saint-Etienne, France
| | - Isabelle Poizot-Martin
- Aix-Marseille Université, APHM, INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, ISSPAM, APHM Sainte-Marguerite, Service d'Immuno-Hématologie Clinique, Marseille, France
| | - Claire Bauduin
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR1219, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | | | - Véronique Rieux
- Vaccine Research Institute, Créteil, France
- INSERM-ANRS, Paris, France
| | - Elodie Rouch
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR1219, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Mathieu Surenaud
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Cécile Lefebvre
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Emile Foucat
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Pascaline Tisserand
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Lydia Guillaumat
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
| | - Mélany Durand
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR1219, Bordeaux, France
- Inria SISTM Team, Talence, France
- Vaccine Research Institute, Créteil, France
| | - Boris Hejblum
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR1219, Bordeaux, France
- Inria SISTM Team, Talence, France
- Vaccine Research Institute, Créteil, France
| | - Odile Launay
- CIC 1417 F-CRIN I-REIVAC, INSERM, Hôpital Cochin, AP-HP, Paris, France; and
- Université Paris Descartes, Paris, France
| | - Rodolphe Thiébaut
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR1219, Bordeaux, France
- Inria SISTM Team, Talence, France
- CHU de Bordeaux, Service d'Information Médicale, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | | |
Collapse
|
110
|
Lin LY, Carapito R, Su B, Moog C. Fc receptors and the diversity of antibody responses to HIV infection and vaccination. Genes Immun 2022; 23:149-156. [PMID: 35688931 PMCID: PMC9388370 DOI: 10.1038/s41435-022-00175-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 11/23/2022]
Abstract
The development of an effective vaccine against HIV is desperately needed. The successive failures of HIV vaccine efficacy trials in recent decades have shown the difficulty of inducing an appropriate protective immune response to fight HIV. Different correlates of antibody parameters associated with a decreased risk of HIV-1 acquisition have been identified. However, these parameters are difficult to reproduce and improve, possibly because they have an intricate and combined action. Here, we describe the numerous antibody (Ab) functions associated with HIV-1 protection and report the interrelated parameters regulating their complex functions. Indeed, besides neutralizing and Fc-mediated activity, additional factors such as Ab type, concentration and kinetics of induction, and Fc-receptor expression and binding capacity also influence the protective effect conferred by Abs. As these parameters were described to be associated with ethnicity, age and sex, these additional factors must be considered for the development of an effective immune response. Therefore, future vaccine designs need to consider these multifaceted Ab functions together with the demographic attributes of the patient populations.
Collapse
Affiliation(s)
- Li-Yun Lin
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Raphael Carapito
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Laboratoire d'Immunologie, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Strasbourg, France
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Christiane Moog
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France. .,Vaccine Research Institute (VRI), Créteil, France.
| |
Collapse
|
111
|
Natural Immunity against HIV-1: Progression of Understanding after Association Studies. Viruses 2022; 14:v14061243. [PMID: 35746714 PMCID: PMC9227919 DOI: 10.3390/v14061243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Abstract
Natural immunity against HIV has been observed in many individuals in the world. Among them, a group of female sex workers enrolled in the Pumwani sex worker cohort remained HIV uninfected for more than 30 years despite high-risk sex work. Many studies have been carried out to understand this natural immunity to HIV in the hope to develop effective vaccines and preventions. This review focuses on two such examples. These studies started from identifying immunogenetic or genetic associations with resistance to HIV acquisition, and followed up with an in-depth investigation to understand the biological relevance of the correlations of protection, and to develop and test novel vaccines and preventions.
Collapse
|
112
|
Harris JE. The repeated setbacks of HIV vaccine development laid the groundwork for SARS-CoV-2 vaccines. HEALTH POLICY AND TECHNOLOGY 2022; 11:100619. [PMID: 35340773 PMCID: PMC8935961 DOI: 10.1016/j.hlpt.2022.100619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The decades-long effort to produce a workable HIV vaccine has hardly been a waste of public and private resources. To the contrary, the scientific know-how acquired along the way has served as the critical foundation for the development of vaccines against the novel, pandemic SARS-CoV-2 virus. We retell the real-world story of HIV vaccine research - with all its false leads and missteps - in a way that sheds light on the current state of the art of antiviral vaccines. We find that HIV-related R&D had more than a general spillover effect. In fact, the repeated failures of phase 2 and 3 clinical trials of HIV vaccine candidates have served as a critical stimulus to the development of successful vaccine technologies today. We rebut the counterargument that HIV vaccine development has been no more than a blind alley, and that recently developed vaccines against COVID-19 are really descendants of successful vaccines against Ebola, MERS, and SARS. These successful vaccines likewise owe much to the vicissitudes of HIV vaccine development. We then discuss how the failures of HIV vaccine development have taught us how adapt SARS-CoV-2 vaccines to immune escape from emerging variants. Finally, we inquire whether recent advances in the development of vaccines against SARS-CoV-2 might in turn further the development of an HIV vaccine - what we describe as a reverse spillover effect.
Collapse
Affiliation(s)
- Jeffrey E Harris
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Eisner Health, Los Angeles, CA 90015, USA
| |
Collapse
|
113
|
Wiysonge CS, Ndwandwe D, Mathebula L, Goga A, Gray G. Randomised trials of COVID-19 vaccines in Africa – charting the path forward. S AFR J SCI 2022. [DOI: 10.17159/sajs.2022/13224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Vaccines have played a critical role in controlling disease outbreaks, hence the proliferation of the development and testing of multiple vaccine candidates during the COVID-19 pandemic. Randomised trials are gold standards for evaluating the safety and efficacy of pharmaceutical interventions such as COVID-19 vaccines. However, contextual differences may attenuate effects of COVID-19 vaccines. Thus, the need to conduct COVID-19 vaccine trials in all settings, including in Africa. We conducted a cross-sectional analysis of planned, ongoing, and completed COVID-19 vaccine trials in Africa. We searched the South African National Clinical Trials Register, Pan African Clinical Trials Registry, and International Clinical Trials Registry Platform (ICTRP) on 12 January and 30 April 2022; and complemented this with a search of ClinicalTrials.gov on 17 May 2022. We screened the search output and included randomised trials with at least one recruitment site in Africa. We identified only 108 eligible trials: 90 (83%) evaluating candidate COVID-19 vaccines, 11 (10%) assessing if existing vaccines could prevent SARS-CoV-2 infection, and 7 (7%) evaluating interventions for improving COVID-19 vaccination coverage. South Africa had the highest number of trials at 58 (54%). Beyond South Africa, countries with more than 10 trial sites include Kenya, Ghana, Egypt, Uganda, and Zimbabwe. Among the trials, 14 (13%) do not have principal investigators based in Africa, 39 (30%) are funded by industry, and 91 (84%) are funded by institutions based outside the host country. COVID-19 vaccine trials with recruitment sites in Africa represented only 7% of the 1453 COVID-19 vaccine trials in the ICTRP. The paucity of COVID-19 vaccine trials conducted on the African continent is a cause for concern. This has implications for the role that Africa may play in future pandemics.
Collapse
Affiliation(s)
- Charles S. Wiysonge
- Cochrane South Africa, South African Medical Research Council, Cape Town, South Africa
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Durban, South Africa
| | - Duduzile Ndwandwe
- Cochrane South Africa, South African Medical Research Council, Cape Town, South Africa
| | - Lindi Mathebula
- Cochrane South Africa, South African Medical Research Council, Cape Town, South Africa
| | - Ameena Goga
- HIV and other Infectious Diseases Research Unit, South African Medical Research Council, Durban, South Africa
- Department of Paediatrics and Child Health, University of Pretoria, Pretoria, South Africa
| | - Glenda Gray
- Office of the President and CEO, South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
114
|
Bernard NF, Alsulami K, Pavey E, Dupuy FP. NK Cells in Protection from HIV Infection. Viruses 2022; 14:v14061143. [PMID: 35746615 PMCID: PMC9231282 DOI: 10.3390/v14061143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 02/05/2023] Open
Abstract
Some people, known as HIV-exposed seronegative (HESN) individuals, remain uninfected despite high levels of exposure to HIV. Understanding the mechanisms underlying their apparent resistance to HIV infection may inform strategies designed to protect against HIV infection. Natural Killer (NK) cells are innate immune cells whose activation state depends on the integration of activating and inhibitory signals arising from cell surface receptors interacting with their ligands on neighboring cells. Inhibitory NK cell receptors use a subset of major histocompatibility (MHC) class I antigens as ligands. This interaction educates NK cells, priming them to respond to cells with reduced MHC class I antigen expression levels as occurs on HIV-infected cells. NK cells can interact with both autologous HIV-infected cells and allogeneic cells bearing MHC antigens seen as non self by educated NK cells. NK cells are rapidly activated upon interacting with HIV-infected or allogenic cells to elicit anti-viral activity that blocks HIV spread to new target cells, suppresses HIV replication, and kills HIV-infected cells before HIV reservoirs can be seeded and infection can be established. In this manuscript, we will review the epidemiological and functional evidence for a role for NK cells in protection from HIV infection.
Collapse
Affiliation(s)
- Nicole F. Bernard
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H4A3J1, Canada; (K.A.); (E.P.); (F.P.D.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Infectious Diseases, Immunology and Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Division of Clinical Immunology, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Correspondence: ; Tel.: +1-(514)-934-1934 (ext. 44584)
| | - Khlood Alsulami
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H4A3J1, Canada; (K.A.); (E.P.); (F.P.D.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Infectious Diseases, Immunology and Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Erik Pavey
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H4A3J1, Canada; (K.A.); (E.P.); (F.P.D.)
- Infectious Diseases, Immunology and Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Franck P. Dupuy
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H4A3J1, Canada; (K.A.); (E.P.); (F.P.D.)
- Infectious Diseases, Immunology and Global Health Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
115
|
Coughlan L, Kremer EJ, Shayakhmetov DM. Adenovirus-based vaccines-a platform for pandemic preparedness against emerging viral pathogens. Mol Ther 2022; 30:1822-1849. [PMID: 35092844 PMCID: PMC8801892 DOI: 10.1016/j.ymthe.2022.01.034] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/24/2022] Open
Abstract
Zoonotic viruses continually pose a pandemic threat. Infection of humans with viruses for which we typically have little or no prior immunity can result in epidemics with high morbidity and mortality. These epidemics can have public health and economic impact and can exacerbate civil unrest or political instability. Changes in human behavior in the past few decades-increased global travel, farming intensification, the exotic animal trade, and the impact of global warming on animal migratory patterns, habitats, and ecosystems-contribute to the increased frequency of cross-species transmission events. Investing in the pre-clinical advancement of vaccine candidates against diverse emerging viral threats is crucial for pandemic preparedness. Replication-defective adenoviral (Ad) vectors have demonstrated their utility as an outbreak-responsive vaccine platform during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Ad vectors are easy to engineer; are amenable to rapid, inexpensive manufacturing; are relatively safe and immunogenic in humans; and, importantly, do not require specialized cold-chain storage, making them an ideal platform for equitable global distribution or stockpiling. In this review, we discuss the progress in applying Ad-based vaccines against emerging viruses and summarize their global safety profile, as reflected by their widespread geographic use during the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS 5535, Montpellier, France.
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Discovery and Developmental Therapeutics Program, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
116
|
Bender Ignacio RA, Long J, Saha A, Nguyen FK, Joudeh L, Valinetz E, Mendelsohn SC, Scriba TJ, Hatherill M, Janes H, Churchyard G, Buchbinder S, Duerr A, Shah JA, Hawn TR. Mycobacterium tuberculosis infection, immune activation, and risk of HIV acquisition. PLoS One 2022; 17:e0267729. [PMID: 35503767 PMCID: PMC9064099 DOI: 10.1371/journal.pone.0267729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Although immune activation is associated with HIV acquisition, the nature of inflammatory profiles that increase HIV risk, which may include responses to M. tuberculosis (Mtb) infection, are not well characterized. METHODS We conducted a nested case-control study using cryopreserved samples from persons who did and did not acquire HIV during the multinational Step clinical trial of the MRKAd5 HIV-1 vaccine. PBMCs from the last HIV-negative sample from incident HIV cases and controls were stimulated with Mtb-specific antigens (ESAT-6/CFP-10) and analyzed by flow cytometry with intracellular cytokine staining and scored with COMPASS. We measured inflammatory profiles with five Correlates of TB Risk (CoR) transcriptomic signatures. Our primary analysis examined the association of latent Mtb infection (LTBI; IFNγ+CD4+ T cell frequency) or RISK6 CoR signature with HIV acquisition. Conditional logistic regression analyses, adjusted for known predictors of HIV acquisition, were employed to assess whether TB-associated immune markers were associated with HIV acquisition. RESULTS Among 465 participants, LTBI prevalence (21.5% controls vs 19.1% cases, p = 0.51) and the RISK6 signature were not higher in those who acquired HIV. In exploratory analyses, Mtb antigen-specific polyfunctional CD4+ T cell COMPASS scores (aOR 0.96, 95% CI 0.77, 1.20) were not higher in those who acquired HIV. Two CoR signatures, Sweeney3 (aOR 1.38 (1.07, 1.78) per SD change) and RESPONSE5 (0.78 (0.61, 0.98)), were associated with HIV acquisition. The transcriptomic pattern used to differentiate active vs latent TB (Sweeney3) was most strongly associated with acquiring HIV. CONCLUSIONS LTBI, Mtb polyfunctional antigen-specific CD4+ T cell activation, and RISK6 were not identified as risks for HIV acquisition. In exploratory transcriptomic analyses, two CoR signatures were associated with HIV risk after adjustment for known behavioral and clinical risk factors. We identified host gene expression signatures associated with HIV acquisition, but the observed effects are likely not mediated through Mtb infection.
Collapse
Affiliation(s)
- Rachel A. Bender Ignacio
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- * E-mail:
| | - Jessica Long
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Aparajita Saha
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Felicia K. Nguyen
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Lara Joudeh
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Ethan Valinetz
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Simon C. Mendelsohn
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Holly Janes
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Gavin Churchyard
- Aurum Institute, Parktown, South Africa
- School of Public Health, University of Witwatersrand, Johannesburg, South Africa
- Department of Medicine, Vanderbilt University, Nashville, TN, United States of America
| | - Susan Buchbinder
- San Francisco Department of Public Health and Departments of Medicine and Epidemiology, University of California San Francisco, San Francisco, CA, United States of America
| | - Ann Duerr
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Javeed A. Shah
- Department of Medicine, University of Washington, Seattle, WA, United States of America
- Veteran Affairs Puget Sound Healthcare System, Seattle, WA, United States of America
| | - Thomas R. Hawn
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
117
|
Klasse PJ, Moore JP. Reappraising the Value of HIV-1 Vaccine Correlates of Protection Analyses. J Virol 2022; 96:e0003422. [PMID: 35384694 PMCID: PMC9044961 DOI: 10.1128/jvi.00034-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2022] [Indexed: 01/09/2023] Open
Abstract
With the much-debated exception of the modestly reduced acquisition reported for the RV144 efficacy trial, HIV-1 vaccines have not protected humans against infection, and a vaccine of similar design to that tested in RV144 was not protective in a later trial, HVTN 702. Similar vaccine regimens have also not consistently protected nonhuman primates (NHPs) against viral acquisition. Conversely, experimental vaccines of different designs have protected macaques from viral challenges but then failed to protect humans, while many other HIV-1 vaccine candidates have not protected NHPs. While efficacy varies more in NHPs than humans, vaccines have failed to protect in the most stringent NHP model. Intense investigations have aimed to identify correlates of protection (CoPs), even in the absence of net protection. Unvaccinated animals and humans vary vastly in their susceptibility to infection and in their innate and adaptive responses to the vaccines; hence, merely statistical associations with factors that do not protect are easily found. Systems biological analyses, including artificial intelligence, have identified numerous candidate CoPs but with no clear consistency within or between species. Proposed CoPs sometimes have only tenuous mechanistic connections to immune protection. In contrast, neutralizing antibodies (NAbs) are a central mechanistic CoP for vaccines that succeed against other viruses, including SARS-CoV-2. No HIV-1 vaccine candidate has yet elicited potent and broadly active NAbs in NHPs or humans, but narrow-specificity NAbs against the HIV-1 isolate corresponding to the immunogen do protect against infection by the autologous virus. Here, we analyze why so many HIV-1 vaccines have failed, summarize the outcomes of vaccination in NHPs and humans, and discuss the value and pitfalls of hunting for CoPs other than NAbs. We contrast the failure to find a consistent CoP for HIV-1 vaccines with the identification of NAbs as the principal CoP for SARS-CoV-2.
Collapse
Affiliation(s)
- P. J. Klasse
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York, USA
| | - John P. Moore
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
118
|
A century of attempts to develop an effective tuberculosis vaccine: Why they failed? Int Immunopharmacol 2022; 109:108791. [PMID: 35487086 DOI: 10.1016/j.intimp.2022.108791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022]
Abstract
Tuberculosis (TB) remains a major global health problem despite widespread use of the Bacillus BCG vaccine. This situation is worsened by co-infection with HIV, and the development of multidrug-resistant Mycobacterium tuberculosis (Mtb) strains. Thus, novel vaccine candidates and improved vaccination strategies are urgently needed in order to reduce the incidence of TB and even to eradicate TB by 2050. Over the last few decades, 23 novel TB vaccines have entered into clinical trials, more than 13 new vaccines have reached various stages of preclinical development, and more than 50 potential candidates are in the discovery stage as next-generation vaccines. Nevertheless, why has a century of attempts to introduce an effective TB vaccine failed? Who should be blamed -scientists, human response, or Mtb strategies? Literature review reveals that the elimination of latent or active Mtb infections in a given population seems to be an epigenetic process. With a better understanding of the connections between bacterial infections and gene expression conditions in epigenetic events, opportunities arise in designing protective vaccines or therapeutic agents, particularly as epigenetic processes can be reversed. Therefore, this review provides a brief overview of different approaches towards novel vaccination strategies and the mechanisms underlying these approaches.
Collapse
|
119
|
Sánchez-Martínez A, Acevedo-Sáenz L, Alzate-Ángel JC, Álvarez CM, Guzmán F, Roman T, Urcuqui-Inchima S, Cardona-Maya WD, Velilla PA. Functional Profile of CD8 + T-Cells in Response to HLA-A*02:01-Restricted Mutated Epitopes Derived from the Gag Protein of Circulating HIV-1 Strains from Medellín, Colombia. Front Immunol 2022; 13:793982. [PMID: 35392101 PMCID: PMC8980466 DOI: 10.3389/fimmu.2022.793982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
CD8+ T-cells play a crucial role in the control of HIV replication. HIV-specific CD8+ T-cell responses rapidly expand since the acute phase of the infection, and it has been observed that HIV controllers harbor CD8+ T-cells with potent anti-HIV capacity. The development of CD8+ T-cell-based vaccine against HIV-1 has focused on searching for immunodominant epitopes. However, the strong immune pressure of CD8+ T-cells causes the selection of viral variants with mutations in immunodominant epitopes. Since HIV-1 mutations are selected under the context of a specific HLA-I, the circulation of viral variants with these mutations is highly predictable based on the most prevalent HLA-I within a population. We previously demonstrated the adaptation of circulating strains of HIV-1 to the HLA-A*02 molecule by identifying mutations under positive selection located in GC9 and SL9 epitopes derived from the Gag protein. Also, we used an in silico prediction approach and evaluated whether the mutations found had a higher or lower affinity to the HLA-A*02. Although this strategy allowed predicting the interaction between mutated peptides and HLA-I, the functional response of CD8+ T-cells that these peptides induce is unknown. In the present work, peripheral blood mononuclear cells from 12 HIV-1+ HLA-A*02:01+ individuals were stimulated with the mutated and wild-type peptides derived from the GC9 and SL9 epitopes. The functional profile of CD8+ T-cells was evaluated using flow cytometry, and the frequency of subpopulations was determined according to their number of functions and the polyfunctionality index. The results suggest that the quality of the response (polyfunctionality) could be associated with the binding affinity of the peptide to the HLA molecule, and the functional profile of specific CD8+ T-cells to mutated epitopes in individuals under cART is maintained.
Collapse
Affiliation(s)
- Alexandra Sánchez-Martínez
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Liliana Acevedo-Sáenz
- Grupo Cuidado Enfermería CES, Facultad de Enfermería, Universidad CES, Medellín, Colombia
| | - Juan Carlos Alzate-Ángel
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia.,Unidad de Micología Médica y Experimental, Corporación para Investigaciones Biológicas, Medellín, Universidad de Santander (CIB-UDES), Bucaramanga, Colombia
| | - Cristian M Álvarez
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Fanny Guzmán
- Núcleo de Biotecnología Curauma, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Tanya Roman
- Núcleo de Biotecnología Curauma, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Walter D Cardona-Maya
- Grupo Reproducción, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Paula Andrea Velilla
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia
| |
Collapse
|
120
|
Logunov DY, Livermore DM, Ornelles DA, Bayer W, Marques E, Czerkinsky C, Dolzhikova IV, Ertl HC. COVID-19 vaccination and HIV-1 acquisition. Lancet 2022; 399:e34-e35. [PMID: 35397866 PMCID: PMC8989395 DOI: 10.1016/s0140-6736(22)00332-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/14/2022] [Indexed: 01/16/2023]
Affiliation(s)
- Denis Y Logunov
- NF Gamaleya National Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | | | - David A Ornelles
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Wibke Bayer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ernesto Marques
- Fundação Oswaldo Cruz - FIOCRUZ, Instituto Aggeu Magalhães-IAM, Department of Virology and Experimental Therapeutics, Recife, Brazil; University of Pittsburgh, School of Public Health, Department of Infectious Diseases, Pittsburgh, PA, USA
| | - Cecil Czerkinsky
- CNRS, INSERM, Institut de Pharmacologie Moleculaire et Cellulaire, Université Nice Sophia Antipolis, Nice, France
| | - Inna V Dolzhikova
- NF Gamaleya National Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | | |
Collapse
|
121
|
Zhang H, He C, Jiang F, Cao S, Zhao B, Ding H, Dong T, Han X, Shang H. A longitudinal analysis of immune escapes from HLA-B*13-restricted T-cell responses at early stage of CRF01_AE subtype HIV-1 infection and implications for vaccine design. BMC Immunol 2022; 23:15. [PMID: 35366796 PMCID: PMC8976269 DOI: 10.1186/s12865-022-00491-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Identifying immunogens which can elicit effective T cell responses against human immunodeficiency virus type 1 (HIV-1) is important for developing a T-cell based vaccine. It has been reported that human leukocyte antigen (HLA)-B*13-restricted T-cell responses contributed to HIV control in subtype B' and C infected individuals. However, the kinetics of B*13-restricted T-cell responses, viral evolution within epitopes, and the impact on disease progression in CRF01_AE subtype HIV-1-infected men who have sex with men (MSM) are not known. RESULTS Interferon-γ ELISPOT assays and deep sequencing of viral RNAs were done in 14 early HLA-B*13-positive CRF01_AE subtype HIV-1-infected MSM. We found that responses to RQEILDLWV (Nef106-114, RV9), GQMREPRGSDI (Gag226-236, GI11), GQDQWTYQI (Pol487-498, GI9), and VQNAQGQMV (Gag135-143, VV9) were dominant. A higher relative magnitude of Gag-specific T-cell responses, contributed to viral control, whereas Nef-specific T-cell responses were associated with rapid disease progression. GI11 (Gag) was conserved and strong GI11 (Gag)-specific T-cell responses showed cross-reactivity with a dominant variant, M228I, found in 3/12 patients; GI11 (Gag)-specific T-cell responses were positively associated with CD4 T-cell counts (R = 0.716, P = 0.046). Interestingly, the GI9 (Pol) epitope was also conserved, but GI9 (Pol)-specific T-cell responses did not influence disease progression (P > 0.05), while a D490G variant identified in one patient did not affect CD4 T-cell counts. All the other epitopes studied [VV9 (Gag), RQYDQILIEI (Pol113-122, RI10), HQSLSPRTL (Gag144-152, HL9), and RQANFLGRL (Gag429-437, RL9)] developed escape mutations within 1 year of infection, which may have contributed to overall disease progression. Intriguingly, we found early RV9 (Nef)-specific T-cell responses were associated with rapid disease progression, likely due to escape mutations. CONCLUSIONS Our study strongly suggested the inclusion of GI11 (Gag) and exclusion of RV9 (Nef) for T-cell-based vaccine design for B*13-positive CRF01_AE subtype HIV-1-infected MSM and high-risk individuals.
Collapse
Affiliation(s)
- Hui Zhang
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| | - Chuan He
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China ,grid.412636.40000 0004 1757 9485Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| | - Fanming Jiang
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China ,grid.412636.40000 0004 1757 9485Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| | - Shuang Cao
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China ,grid.412449.e0000 0000 9678 1884Department of Laboratory Medicine, China Medical University Shengjing Hospital Nanhu Branch, Shenyang, 110001 China
| | - Bin Zhao
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| | - Haibo Ding
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| | - Tao Dong
- grid.4991.50000 0004 1936 8948Nuffield Department of Medicine, Chinese Academy of Medical Sciences Oxford Institute, Oxford University, Oxford, UK ,grid.4991.50000 0004 1936 8948Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK
| | - Xiaoxu Han
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| | - Hong Shang
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| |
Collapse
|
122
|
Nkone P, Loubser S, Quinn TC, Redd AD, Ismail A, Tiemessen CT, Mayaphi SH. Deep sequencing of the HIV-1 polymerase gene for characterisation of cytotoxic T-lymphocyte epitopes during early and chronic disease stages. Virol J 2022; 19:56. [PMID: 35346259 PMCID: PMC8959563 DOI: 10.1186/s12985-022-01772-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/07/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Despite multiple attempts, there is still no effective HIV-1 vaccine available. The HIV-1 polymerase (pol) gene is highly conserved and encodes cytotoxic T-lymphocyte (CTL) epitopes. The aim of the study was to characterise HIV-1 Pol CTL epitopes in mostly sample pairs obtained during early and chronic stages of infection. METHODS Illumina deep sequencing was performed for all samples while Sanger sequencing was only performed on baseline samples. Codons under immune selection pressure were assessed by computing nonsynonymous to synonymous mutation ratios using MEGA. Minority CTL epitope variants occurring at [Formula: see text] 5% were detected using low-frequency variant tool in CLC Genomics. Los Alamos HIV database was used for mapping mutations to known HIV-1 CTL epitopes. RESULTS Fifty-two participants were enrolled in the study. Their median age was 28 years (interquartile range: 24-32 years) and majority of participants (92.3%) were female. Illumina minority variant analysis identified a significantly higher number of CTL epitopes (n = 65) compared to epitopes (n = 8) identified through Sanger sequencing. Most of the identified epitopes mapped to reverse transcriptase (RT) and integrase (IN) regardless of sequencing method. There was a significantly higher proportion of minority variant epitopes in RT (n = 39, 60.0%) compared to IN (n = 17, 26.2%) and PR (n = 9, 13.8%), p = 0.002 and < 0.0001, respectively. However, no significant difference was observed between the proportion of minority variant epitopes in IN versus PR, p = 0.06. Some epitopes were detected in either early or chronic HIV-1 infection whereas others were detected in both stages. Different distribution patterns of minority variant epitopes were observed in sample pairs; with some increasing or decreasing over time, while others remained constant. Some of the identified epitopes have not been previously reported for HIV-1 subtype C. There were also variants that could not be mapped to reported CTL epitopes in the Los Alamos HIV database. CONCLUSION Deep sequencing revealed many Pol CTL epitopes, including some not previously reported for HIV-1 subtype C. The findings of this study support the inclusion of RT and IN epitopes in HIV-1 vaccine candidates as these proteins harbour many CTL epitopes.
Collapse
Affiliation(s)
- Paballo Nkone
- Department of Medical Virology, University of Pretoria, Private Bag X323, Gezina, 0031, South Africa
| | - Shayne Loubser
- National Institute for Communicable Diseases and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Thomas C Quinn
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew D Redd
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Arshad Ismail
- National Institute for Communicable Diseases and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Caroline T Tiemessen
- National Institute for Communicable Diseases and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Simnikiwe H Mayaphi
- Department of Medical Virology, University of Pretoria, Private Bag X323, Gezina, 0031, South Africa. .,National Health Laboratory Service-Tshwane Academic Division (NHLS-TAD), Tshwane, South Africa.
| |
Collapse
|
123
|
Liu Y, Ye Q. Safety and Efficacy of the Common Vaccines against COVID-19. Vaccines (Basel) 2022; 10:vaccines10040513. [PMID: 35455262 PMCID: PMC9027683 DOI: 10.3390/vaccines10040513] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023] Open
Abstract
The worldwide pandemic of coronavirus disease 2019 (COVID-19) has imposed a challenge on human health worldwide, and vaccination represents a vital strategy to control the pandemic. To date, multiple COVID-19 vaccines have been granted emergency use authorization, including inactivated vaccines, adenovirus-vectored vaccines, and nucleic acid vaccines. These vaccines have different technical principles, which will necessarily lead to differences in safety and efficacy. Therefore, we aim to implement a systematic review by synthesizing clinical experimental data combined with mass vaccination data and conducting a synthesis to evaluate the safety and efficacy of COVID-19 vaccines. Compared with other vaccines, adverse reactions after vaccination with inactivated vaccines are relatively low. The efficacy of inactivated vaccines is approximately 60%, adenovirus-vectored vaccines are 65%, and mRNA vaccines are 90%, which are always efficient against asymptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, symptomatic COVID-19, COVID-19 hospitalization, severe or critical hospitalization, and death. RNA-based vaccines have a number of advantages and are one of the most promising vaccines identified to date and are particularly important during a pandemic. However, further improvements are required. In time, all the antibody levels weaken gradually, so a booster dose is needed to maintain immunity. Compared with homologous prime-boost immunization, heterologous prime-boost immunization prompts more robust humoral and cellular immune responses.
Collapse
|
124
|
Cytokine Adjuvants IL-7 and IL-15 Improve Humoral Responses of a SHIV LentiDNA Vaccine in Animal Models. Vaccines (Basel) 2022; 10:vaccines10030461. [PMID: 35335093 PMCID: PMC8949948 DOI: 10.3390/vaccines10030461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/02/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023] Open
Abstract
HIV-1 remains a major public health issue worldwide in spite of efficacious antiviral therapies, but with no cure or preventive vaccine. The latter has been very challenging, as virus infection is associated with numerous escape mechanisms from host specific immunity and the correlates of protection remain incompletely understood. We have developed an innovative vaccine strategy, inspired by the efficacy of live-attenuated virus, but with the safety of a DNA vaccine, to confer both cellular and humoral responses. The CAL-SHIV-IN− lentiDNA vaccine comprises the backbone of the pathogenic SHIVKU2 genome, able to mimic the early phase of viral infection, but with a deleted integrase gene to ensure safety precluding integration within the host genome. This vaccine prototype, constitutively expressing viral antigen under the CAEV LTR promoter, elicited a variety of vaccine-specific, persistent CD4 and CD8 T cells against SIV-Gag and Nef up to 80 weeks post-immunization in cynomolgus macaques. Furthermore, these specific responses led to antiviral control of the pathogenic SIVmac251. To further improve the efficacy of this vaccine, we incorporated the IL-7 or IL-15 genes into the CAL-SHIV-IN− plasmid DNA in efforts to increase the pool of vaccine-specific memory T cells. In this study, we examined the immunogenicity of the two co-injected lentiDNA vaccines CAL-SHIV-IN− IRES IL-7 and CAL-SHIV-IN− IRES IL-15 in BALB/cJ mice and rhesus macaques and compared the immune responses with those generated by the parental vaccine CAL-SHIV-IN−. This co-immunization elicited potent vaccine-specific CD4 and CD8 T cells both in mice and rhesus macaques. Antibody-dependent cell-mediated cytotoxicity (ADCC) antibodies were detected up to 40 weeks post-immunization in both plasma and mucosal compartments of rhesus macaques and were enhanced by the cytokines.
Collapse
|
125
|
Enhanced Cross-Reactive and Polyfunctional Effector-Memory T Cell Responses by ICVAX-a Human PD1-Based Bivalent HIV-1 Gag-p41 Mosaic DNA Vaccine. J Virol 2022; 96:e0216121. [PMID: 35297660 PMCID: PMC9006887 DOI: 10.1128/jvi.02161-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vaccine-induced protective T cell immunity is necessary for HIV-1 functional cure. We previously reported that rhesus PD1-Gag-based DNA vaccination sustained simian-human immunodeficiency virus (SHIV) suppression by inducing effector-memory CD8+ T cells. Here, we investigated a human PD1-Gag-based DNA vaccine, namely, ICVAX, for clinical translation. PD1-based dendritic cell targeting and mosaic antigenic designs were combined to generate the ICVAX by fusing the human soluble PD1 domain with a bivalent HIV-1 Gag-p41 mosaic antigen. The mosaic antigen was cross-reactive with patients infected with B, CRF07/08_BC, and CRF01_AE variants. In mice, ICVAX elicited stronger, broader, and more polyfunctional T cell responses than mosaic Gag-p41 alone, and suppressed EcoHIV infection more efficiently. In macaques, ICVAX elicited polyfunctional effector-memory T cell responses that targeted multiple nonoverlapping epitopes of the Gag-p41 antigen. Furthermore, ICVAX manufactured following good manufacturing practices proved potent immunogenicity in macaques after biannual homologous vaccination, warranting clinical evaluation of ICVAX as an immunotherapy against HIV-1. IMPORTANCE This study presents that ICVAX, a PD1-based DNA vaccine against HIV-1, could induce broad and polyfunctional T cell responses against different HIV-1 subtypes. ICVAX encodes a recombinant antigen consisting of the human soluble PD1 domain fused with two mosaic Gag-p41 antigens. The mosaic antigens cover more than 500 HIV-1 strains circulating in China including the subtypes B/B’, CRF01_AE, and CRF07/08_BC. In mice, ICVAX elicited stronger, broader, and more polyfunctional T cell responses, with better EcoHIV suppression than the nontargeting mosaic Gag-p41 DNA vaccine. Moreover, both lab-generated and GMP-grade ICVAX also elicited strong polyfunctional effector-memory T cell responses in rhesus macaques with good immunogenicity against multiple nonoverlapping epitopes of the Gag-p41 antigen. This study therefore highlights the great potential to translate the PD1-based DNA vaccine approach into clinical use, and opens up new avenues for alternative HIV-1 vaccine design for HIV-1 preventive and functional cure.
Collapse
|
126
|
Guo X, Sun Y, Chen J, Zou X, Hou W, Tan W, Hung T, Lu Z. Restriction-Assembly: A Solution to Construct Novel Adenovirus Vector. Viruses 2022; 14:v14030546. [PMID: 35336953 PMCID: PMC8954691 DOI: 10.3390/v14030546] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 01/27/2023] Open
Abstract
Gene therapy and vaccine development need more novel adenovirus vectors. Here, we attempt to provide strategies to construct adenovirus vectors based on restriction-assembly for researchers with little experience in this field. Restriction-assembly is a combined method of restriction digestion and Gibson assembly, by which the major part of the obtained plasmid comes from digested DNA fragments instead of PCR products. We demonstrated the capability of restriction-assembly in manipulating the genome of simian adenovirus 1 (SAdV-1) in this study. A PCR product of the plasmid backbone was combined with SAdV-1 genomic DNA to construct an infectious clone, plasmid pKSAV1, by Gibson assembly. Restriction-assembly was performed repeatedly in the steps of intermediate plasmid isolation, modification, and restoration. The generated adenoviral plasmid was linearized by restriction enzyme digestion and transfected into packaging 293 cells to rescue E3-deleted replication-competent SAdV1XE3-CGA virus. Interestingly, SAdV1XE3-CGA could propagate in human chronic myelogenous leukemia K562 cells. The E1 region was similarly modified to generate E1/E3-deleted replication-defective virus SAdV1-EG. SAdV1-EG had a moderate gene transfer ability to adherent mammalian cells, and it could efficiently transduce suspension cells when compared with the human adenovirus 5 control vector. Restriction-assembly is easy to use and can be performed without special experimental materials and instruments. It is highly effective with verifiable outcomes at each step. More importantly, restriction-assembly makes the established vector system modifiable, upgradable and under sustainable development, and it can serve as the instructive method or strategy for the synthetic biology of adenoviruses.
Collapse
Affiliation(s)
- Xiaojuan Guo
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (X.G.); (Y.S.); (J.C.); (X.Z.); (W.H.); (T.H.)
| | - Yangyang Sun
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (X.G.); (Y.S.); (J.C.); (X.Z.); (W.H.); (T.H.)
- School of Laboratory Medicine, Weifang Medical University, Weifang 261053, China
| | - Juan Chen
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (X.G.); (Y.S.); (J.C.); (X.Z.); (W.H.); (T.H.)
- School of Public Health, Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Xiaohui Zou
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (X.G.); (Y.S.); (J.C.); (X.Z.); (W.H.); (T.H.)
| | - Wenzhe Hou
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (X.G.); (Y.S.); (J.C.); (X.Z.); (W.H.); (T.H.)
| | - Wenjie Tan
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (X.G.); (Y.S.); (J.C.); (X.Z.); (W.H.); (T.H.)
- Correspondence: (Z.L.); (W.T.); Tel.: +86-10-63511368 (Z.L.)
| | - Tao Hung
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (X.G.); (Y.S.); (J.C.); (X.Z.); (W.H.); (T.H.)
| | - Zhuozhuang Lu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 100052, China; (X.G.); (Y.S.); (J.C.); (X.Z.); (W.H.); (T.H.)
- Chinese Center for Disease Control and Prevention–Wuhan Institute of Virology, Chinese Academy of Sciences Joint Research Center for Emerging Infectious Diseases and Biosafety, Wuhan 430071, China
- Correspondence: (Z.L.); (W.T.); Tel.: +86-10-63511368 (Z.L.)
| |
Collapse
|
127
|
Toh CH, Wang G, Parker AL. The aetiopathogenesis of vaccine-induced immune thrombotic thrombocytopenia. Clin Med (Lond) 2022; 22:140-144. [PMID: 38589175 PMCID: PMC8966812 DOI: 10.7861/clinmed.2022-0006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In the new science emanating from the COVID-19 pandemic, effective vaccine development has made a huge difference and saved countless lives. Vaccine roll-out led to the identification of rare cases of severe thrombotic and thrombocytopenic problems in some recipients. This apparent coupling of thrombosis with haemorrhagic potentiation might seem baffling but the ensuing clinical investigation rapidly shed important light on its molecular mechanism. This review outlines the current understanding on the role of adenovirus-based platforms, the immunogenic triggers and the immunothrombotic response underlying vaccine-induced immune thrombotic thrombocytopenia.
Collapse
|
128
|
Abstract
This review discusses peptide epitopes used as antigens in the development of vaccines in clinical trials as well as future vaccine candidates. It covers peptides used in potential immunotherapies for infectious diseases including SARS-CoV-2, influenza, hepatitis B and C, HIV, malaria, and others. In addition, peptides for cancer vaccines that target examples of overexpressed proteins are summarized, including human epidermal growth factor receptor 2 (HER-2), mucin 1 (MUC1), folate receptor, and others. The uses of peptides to target cancers caused by infective agents, for example, cervical cancer caused by human papilloma virus (HPV), are also discussed. This review also provides an overview of model peptide epitopes used to stimulate non-specific immune responses, and of self-adjuvanting peptides, as well as the influence of other adjuvants on peptide formulations. As highlighted in this review, several peptide immunotherapies are in advanced clinical trials as vaccines, and there is great potential for future therapies due the specificity of the response that can be achieved using peptide epitopes.
Collapse
Affiliation(s)
- Ian W Hamley
- Department of Chemistry, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| |
Collapse
|
129
|
Jeewandara C, Fernando S, Pushpakumara PD, Ramu ST, Kamaladasa A, Gunasekara B, Aberathna IS, Kuruppu H, Ranasinghe T, Dayarathne S, Dissanayake O, Gamalath N, Ekanayake D, Jayamali J, Wijesinghe A, Dissanayake M, Somathilake G, Harvie M, Danasekara S, Jayathilaka D, Wijayatilake HDK, Weerasooriya N, Kekulandara C, Schimanski L, Rijal P, Tan TK, Dong T, Townsend A, Ogg GS, Malavige GN. Immune responses following the first dose of the Sputnik V (Gam-COVID-Vac). Sci Rep 2022; 12:1727. [PMID: 35110645 PMCID: PMC8810924 DOI: 10.1038/s41598-022-05788-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 01/12/2022] [Indexed: 01/08/2023] Open
Abstract
As the first dose of Gam-COVID-Vac, is currently used as a single dose vaccine in some countries, we investigated the immunogenicity of this at 4 weeks (327 naïve individuals). 88.7% seroconverted, with significantly lower seroconversion rates in those over 60 years (p = 0.004) and significantly lower than previously seen with AZD1222 (p = 0.018). 82.6% developed ACE2 receptor blocking antibodies, although levels were significantly lower than following natural infection (p = 0.0009) and a single dose of AZD1222 (p < 0.0001). Similar titres of antibodies were observed to the receptor binding domain of WT, B.1.1.7 and B.1.617.2 compared to AZD1222, while the levels for B.1.351 were significantly higher (p = 0.006) for Gam-COVID-Vac. 30% developed ex vivo IFNγ ELISpot responses (significantly lower than AZD1222), and high frequency of CD107a expressing T cells along with memory B cell responses. Although single dose of Gam-COVID-Vac was highly immunogenic, administration of a second dose is likely to be beneficial.
Collapse
Affiliation(s)
- Chandima Jeewandara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | | | - Pradeep Darshana Pushpakumara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Shyrar Tanussiya Ramu
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Achala Kamaladasa
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Banuri Gunasekara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Inoka Sepali Aberathna
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Heshan Kuruppu
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Thushali Ranasinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Shashika Dayarathne
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Osanda Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Nayanathara Gamalath
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Dinithi Ekanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Jewantha Jayamali
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Ayesha Wijesinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Madushika Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Gayasha Somathilake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Michael Harvie
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Saubhagya Danasekara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | - Deshni Jayathilaka
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka
| | | | | | | | - Lisa Schimanski
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Pramila Rijal
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Tiong K Tan
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Tao Dong
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Alain Townsend
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Graham S Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Centre for Translational Immunology, Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Gathsaurie Neelika Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayawardanapura, Nugegoda, Sri Lanka.
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
130
|
Nomah DK, Reyes-Urueña J, Llibre JM, Ambrosioni J, Ganem FS, Miró JM, Casabona J. HIV and SARS-CoV-2 Co-infection: Epidemiological, Clinical Features, and Future Implications for Clinical Care and Public Health for People Living with HIV (PLWH) and HIV Most-at-Risk Groups. Curr HIV/AIDS Rep 2022; 19:17-25. [PMID: 35113346 PMCID: PMC8810339 DOI: 10.1007/s11904-021-00596-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to use the currently available clinical and epidemiological data, to identify key aspects to improve both the clinical management and public health response to SARS-CoV-2/HIV co-infection among HIV vulnerable populations and people living with HIV (PLWH). RECENT FINDINGS While at the beginning of the COVID-19 pandemic, the lack of robust information on SARS-CoV-2/HIV co-infection, prevented a clear picture of the synergies between them, currently available data strongly support the importance of common structural factors on both the acquisition and clinical impact of these infections and the relevance of age, comorbidities, and detectable HIV viral load as associated worse prognostic factors among PLWH. Although more information is needed to better understand the biological, clinical, and epidemiological relationship between both infections, a syndemic approach to prevent SARS-CoV-2 among HIV high-risk groups and PLWH, targeting these populations for SARS-CoV-2 vaccines and protocolizing early identification of PLWH with worse COVID-19 prognosis factors, is crucial strategies to decrease the overall impact of SARS-CoV-2 /HIV co-infection.
Collapse
Affiliation(s)
- D K Nomah
- Centre Estudis Epidemiològics sobre les Infeccions de Transmissió Sexual i Sida de Catalunya (CEEISCAT), ICO/Departament de Salut, Generalitat de Catalunya, Badalona, Spain
- Departament de Pediatria, d'Obstetrícia i Ginecologia i de Medicina Preventiva i de Salut Publica, Universitat Autònoma de Barcelona, Bellaterra, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain
| | - J Reyes-Urueña
- Centre Estudis Epidemiològics sobre les Infeccions de Transmissió Sexual i Sida de Catalunya (CEEISCAT), ICO/Departament de Salut, Generalitat de Catalunya, Badalona, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain
- Institut d''Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - J M Llibre
- Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - J Ambrosioni
- Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - F S Ganem
- Centre Estudis Epidemiològics sobre les Infeccions de Transmissió Sexual i Sida de Catalunya (CEEISCAT), ICO/Departament de Salut, Generalitat de Catalunya, Badalona, Spain
- Departament de Pediatria, d'Obstetrícia i Ginecologia i de Medicina Preventiva i de Salut Publica, Universitat Autònoma de Barcelona, Bellaterra, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain
| | - J M Miró
- Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - J Casabona
- Centre Estudis Epidemiològics sobre les Infeccions de Transmissió Sexual i Sida de Catalunya (CEEISCAT), ICO/Departament de Salut, Generalitat de Catalunya, Badalona, Spain.
- Departament de Pediatria, d'Obstetrícia i Ginecologia i de Medicina Preventiva i de Salut Publica, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain.
- Institut d''Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Barcelona, Spain.
| |
Collapse
|
131
|
Sakurai F, Tachibana M, Mizuguchi H. Adenovirus vector-based vaccine for infectious diseases. Drug Metab Pharmacokinet 2022; 42:100432. [PMID: 34974335 PMCID: PMC8585960 DOI: 10.1016/j.dmpk.2021.100432] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 01/10/2023]
Abstract
Replication-incompetent adenovirus (Ad) vectors have been widely used as gene delivery vehicles in both gene therapy studies and basic studies for gene function analysis due to their highly advantageous properties, which include high transduction efficiencies, relatively large capacities for transgenes, and high titer production. In addition, Ad vectors induce moderate levels of innate immunity and have relatively high thermostability, making them very attractive as potential vaccine vectors. Accordingly, it is anticipated that Ad vectors will be used in vaccines for the prevention of infectious diseases, including Ebola virus disease and acquired immune deficiency syndrome (AIDS). Much attention is currently focused on the potential use of an Ad vector vaccine for coronavirus disease 2019 (COVID-19). In this review, we describe the basic properties of an Ad vector, Ad vector-induced innate immunity and immune responses to Ad vector-produced transgene products. Development of novel Ad vectors which can overcome the drawbacks of conventional Ad vector vaccines and clinical application of Ad vector vaccines to several infectious diseases are also discussed.
Collapse
Affiliation(s)
- Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
| | - Masashi Tachibana
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan; Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
| |
Collapse
|
132
|
Pavia CS, Plummer MM. COVID-19 vaccines for high risk and immunocompromised patients. METHODS IN MICROBIOLOGY 2022; 50:269-279. [PMID: 38620783 PMCID: PMC8797165 DOI: 10.1016/bs.mim.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The availability of multiple versions of vaccines designed to help prevent COVID-19 has offered an opportunity to at least control the current pandemic, and possibly to quickly eradicate this disease fully, along with the implementation of other preventive measures. In order to accomplish this feat more effectively, as many people as possible need to be vaccinated, especially for high-risk groups having co-morbid conditions such as diabetes, obesity and old age, and possibly those with various forms of immunodeficiencies, such as HIV/AIDS. This chapter focuses primarily on some of the basic biomedical aspects on vaccine design and use, and any possible concerns that need to be considered in getting people in the high-risk category vaccinated and monitored thereafter for their continuous health and well-being.
Collapse
Affiliation(s)
- Charles S Pavia
- Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, United States
- Division of Infectious Diseases, New York Medical College, Valhalla, NY, United States
| | - Maria M Plummer
- Department of Clinical Specialties, Division of Pathology, NYIT College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, United States
| |
Collapse
|
133
|
Herrera C, Veazey R, Lemke MM, Arnold K, Kim JH, Shattock RJ. Ex Vivo Evaluation of Mucosal Responses to Vaccination with ALVAC and AIDSVAX of Non-Human Primates. Vaccines (Basel) 2022; 10:187. [PMID: 35214645 PMCID: PMC8879115 DOI: 10.3390/vaccines10020187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 02/01/2023] Open
Abstract
Non-human primates (NHPs) remain the most relevant challenge model for the evaluation of HIV vaccine candidates; however, discrepancies with clinical trial results have emphasized the need to further refine the NHP model. Furthermore, classical evaluation of vaccine candidates is based on endpoints measured systemically. We assessed the mucosal responses elicited upon vaccination with ALVAC and AIDSVAX using ex vivo Rhesus macaque mucosal tissue explant models. Following booster immunization with ALVAC/AIDSVAX, anti-gp120 HIV-1CM244-specific IgG and IgA were detected in culture supernatant cervicovaginal and colorectal tissue explants, as well as systemically. Despite protection from ex vivo viral challenge, no neutralization was observed with tissue explant culture supernatants. Priming with ALVAC induced distinct cytokine profiles in cervical and rectal tissue. However, ALVAC/AIDSVAX boosts resulted in similar modulations in both mucosal tissues with a statistically significant decrease in cytokines linked to inflammatory responses and lymphocyte differentiation. With ALVAC/AIDSVAX boosts, significant correlations were observed between cytokine levels and specific IgA in cervical explants and specific IgG and IgA in rectal tissue. The cytokine secretome revealed differences between vaccination with ALVAC and ALVAC/AIDSVAX not previously observed in mucosal tissues and distinct from the systemic response, which could represent a biosignature of the vaccine combination.
Collapse
Affiliation(s)
- Carolina Herrera
- Department of Medicine, Imperial College London, London W2 1PG, UK;
| | - Ronald Veazey
- Tulane National Primate Research Center, School of Medicine, Tulane University, Covington, GA 70433, USA;
| | - Melissa M. Lemke
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (M.M.L.); (K.A.)
| | - Kelly Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (M.M.L.); (K.A.)
| | - Jerome H. Kim
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MA 20817, USA;
| | | |
Collapse
|
134
|
Roth GA, Picece VCTM, Ou BS, Luo W, Pulendran B, Appel EA. Designing spatial and temporal control of vaccine responses. NATURE REVIEWS. MATERIALS 2022; 7:174-195. [PMID: 34603749 PMCID: PMC8477997 DOI: 10.1038/s41578-021-00372-2] [Citation(s) in RCA: 174] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 05/02/2023]
Abstract
Vaccines are the key technology to combat existing and emerging infectious diseases. However, increasing the potency, quality and durability of the vaccine response remains a challenge. As our knowledge of the immune system deepens, it becomes clear that vaccine components must be in the right place at the right time to orchestrate a potent and durable response. Material platforms, such as nanoparticles, hydrogels and microneedles, can be engineered to spatially and temporally control the interactions of vaccine components with immune cells. Materials-based vaccination strategies can augment the immune response by improving innate immune cell activation, creating local inflammatory niches, targeting lymph node delivery and controlling the time frame of vaccine delivery, with the goal of inducing enhanced memory immunity to protect against future infections. In this Review, we highlight the biological mechanisms underlying strong humoral and cell-mediated immune responses and explore materials design strategies to manipulate and control these mechanisms.
Collapse
Affiliation(s)
- Gillie A. Roth
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Vittoria C. T. M. Picece
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Ben S. Ou
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Wei Luo
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA USA
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford, CA USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Paediatrics — Endocrinology, Stanford University School of Medicine, Stanford, CA USA
| |
Collapse
|
135
|
Abstract
The arrival of the most recent coronavirus in 2019, SARS-CoV-2, caught the entire world by surprise, and as a result has caused more anguish due to its rapid spread and serious health consequences for the elderly and those with underlying health conditions, and its ability to generate variants of ever increasing contagiousness. But this was not the first coronavirus to infect humans. This chapter explores the history of this virus family, the emergence of the first serious infection in 2003–04 (SARS-CoV), and the related virus MERS in 2012, and the possible origins of SARS-CoV-2. The lessons of those two outbreaks that never developed into pandemics may not all have been learnt by the world health leaders of today. Nevertheless, the rapidity of vaccine development and the conventional health measure introduced during 2020, not always in good time, has almost certainly led to lower morbidities and mortalities that would otherwise have been the case. This chapter will inevitably be out of date by time this book goes to press. Nevertheless, it is to be hoped that the origin of SARS-CoV-2 will eventually be established, but sadly not without the cooperation of the major countries having the resources to carry out such complex investigations. If such a cooperation did happen, maybe future pandemics of this will be more controllable, and even never progress beyond local outbreaks.
Collapse
|
136
|
Barin F, Stefic K. [Protection against HIV-1 infection: "It's the neutralizing antibody, stupid!"]. Med Sci (Paris) 2021; 37:1178-1179. [PMID: 34928225 DOI: 10.1051/medsci/2021175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Francis Barin
- Inserm U1259, université de Tours, 10 boulevard Tonnellé, 37004 Tours, France
| | - Karl Stefic
- Inserm U1259, université de Tours, 10 boulevard Tonnellé, 37004 Tours, France - Laboratoire de virologie et CNR VIH-laboratoire associé, CHU Bretonneau, 2 boulevard Tonnellé, 37004 Tours, France
| |
Collapse
|
137
|
Lassaunière R, Tiemessen CT. FcγR Genetic Variation and HIV-1 Vaccine Efficacy: Context And Considerations. Front Immunol 2021; 12:788203. [PMID: 34975881 PMCID: PMC8714752 DOI: 10.3389/fimmu.2021.788203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/29/2021] [Indexed: 12/02/2022] Open
Abstract
Receptors for the crystallisable fragment (Fc) of immunoglobulin (Ig) G, Fcγ receptors (FcγRs), link the humoral and cellular arms of the immune response, providing a diverse armamentarium of antimicrobial effector functions. Findings from HIV-1 vaccine efficacy trials highlight the need for further study of Fc-FcR interactions in understanding what may constitute vaccine-induced protective immunity. These include host genetic correlates identified within the low affinity Fcγ-receptor locus in three HIV-1 efficacy trials – VAX004, RV144, and HVTN 505. This perspective summarizes our present knowledge of FcγR genetics in the context of findings from HIV-1 efficacy trials, and draws on genetic variation described in other contexts, such as mother-to-child HIV-1 transmission and HIV-1 disease progression, to explore the potential contribution of FcγR variability in modulating different HIV-1 vaccine efficacy outcomes. Appreciating the complexity and the importance of the collective contribution of variation within the FCGR gene locus is important for understanding the role of FcγRs in protection against HIV-1 acquisition.
Collapse
Affiliation(s)
- Ria Lassaunière
- Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
- *Correspondence: Caroline T. Tiemessen, ; Ria Lassaunière,
| | - Caroline T. Tiemessen
- Centre for HIV and STI’s, National Institute for Communicable Diseases, Johannesburg, South Africa
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- *Correspondence: Caroline T. Tiemessen, ; Ria Lassaunière,
| |
Collapse
|
138
|
Tsakiri M, Naziris N, Demetzos C. Innovative vaccine platforms against infectious diseases: Under the scope of the COVID-19 pandemic. Int J Pharm 2021; 610:121212. [PMID: 34687816 PMCID: PMC8527590 DOI: 10.1016/j.ijpharm.2021.121212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/06/2021] [Accepted: 10/15/2021] [Indexed: 12/30/2022]
Abstract
While classic vaccines have proved greatly efficacious in eliminating serious infectious diseases, innovative vaccine platforms open a new pathway to overcome dangerous pandemics via the development of safe and effective formulations. Such platforms play a key role either as antigen delivery systems or as immune-stimulators that induce both innate and adaptive immune responses. Liposomes or lipid nanoparticles, virus-like particles, nanoemulsions, polymeric or inorganic nanoparticles, as well as viral vectors, all belong to the nanoscale and are the main categories of innovative vaccines that are currently on the market or in clinical and preclinical phases. In this paper, we review the above formulations used in vaccinology and we discuss their connection with the development of safe and effective prophylactic vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Maria Tsakiri
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Nikolaos Naziris
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece.
| |
Collapse
|
139
|
Sanchez S, Palacio N, Dangi T, Ciucci T, Penaloza-MacMaster P. Fractionating a COVID-19 Ad5-vectored vaccine improves virus-specific immunity. Sci Immunol 2021; 6:eabi8635. [PMID: 34648369 DOI: 10.1126/sciimmunol.abi8635] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nicole Palacio
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Thomas Ciucci
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA.,Department of Microbiology and Immunology, Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
140
|
Nomah DK, Reyes-Urueña J, Llibre JM, Ambrosioni J, Ganem FS, Miró JM, Casabona J. HIV and SARS-CoV-2 Co-infection: Epidemiological, Clinical Features, and Future Implications for Clinical Care and Public Health for People Living with HIV (PLWH) and HIV Most-at-Risk Groups. Curr HIV/AIDS Rep 2021; 18:518-526. [PMID: 34890019 PMCID: PMC8661831 DOI: 10.1007/s11904-021-00579-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is using the currently available clinical and epidemiological data, to identify key aspects to improve both the clinical management and public health response with regard SARS-CoV-2/HIV co-infection among HIV vulnerable populations and people living with HIV (PLWH). RECENT FINDINGS While at the beginning of the COVID-19 pandemic, the lack of robust information on SARS-CoV-2/HIV coinfection prevented to have a clear picture of the synergies between them, currently available data strongly supports the importance of common structural factors on both the acquisition and clinical impact of these infections and the relevance of age, co-morbidities, and HIV viral load as associated worse prognosis factors among PLWH. Although more information is needed to better understand the biological, clinical, and epidemiological relationship between both infections, in the meanwhile, syndemic approaches to prevent SARS-CoV-2 among HIV higher risk groups and PLWH, targeting these population for SARS-CoV-2 vaccines and protocolizing early identification of HIV + patients with worse COVID-19 prognosis factors, are crucial strategies to decrease the overall impact of SARS-CoV-2 /HIV coinfection.
Collapse
Affiliation(s)
- Daniel K Nomah
- Centre Estudis Epidemiològics Sobre Les Infeccions de Transmissió Sexual I Sida de Catalunya (CEEISCAT), Departament de Salut, Generalitat de Catalunya, Campus de Can Ruti, Josep Carreras Building, Ctra de Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Catalonia, Spain
- Departament de Pediatria, d'Obstetrícia I Ginecologia I de Medicina Preventiva I de Salut Publica, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Institut d'Investigació Germans Trias I Pujol (IGTP), Badalona, Spain
| | - Juliana Reyes-Urueña
- Centre Estudis Epidemiològics Sobre Les Infeccions de Transmissió Sexual I Sida de Catalunya (CEEISCAT), Departament de Salut, Generalitat de Catalunya, Campus de Can Ruti, Josep Carreras Building, Ctra de Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Catalonia, Spain
- Institut d'Investigació Germans Trias I Pujol (IGTP), Badalona, Spain
- CIBER Epidemiologia Y Salud Pública (CIBERESP), Barcelona, Spain
| | - Josep Ma Llibre
- Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | - Juan Ambrosioni
- Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Fabiana S Ganem
- Centre Estudis Epidemiològics Sobre Les Infeccions de Transmissió Sexual I Sida de Catalunya (CEEISCAT), Departament de Salut, Generalitat de Catalunya, Campus de Can Ruti, Josep Carreras Building, Ctra de Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Catalonia, Spain
- Departament de Pediatria, d'Obstetrícia I Ginecologia I de Medicina Preventiva I de Salut Publica, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Institut d'Investigació Germans Trias I Pujol (IGTP), Badalona, Spain
| | - José Ma Miró
- Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer, University of Barcelona, Barcelona, Spain
| | - Jordi Casabona
- Centre Estudis Epidemiològics Sobre Les Infeccions de Transmissió Sexual I Sida de Catalunya (CEEISCAT), Departament de Salut, Generalitat de Catalunya, Campus de Can Ruti, Josep Carreras Building, Ctra de Can Ruti, Camí de les Escoles, s/n, 08916, Badalona, Catalonia, Spain.
- Departament de Pediatria, d'Obstetrícia I Ginecologia I de Medicina Preventiva I de Salut Publica, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- Institut d'Investigació Germans Trias I Pujol (IGTP), Badalona, Spain.
- CIBER Epidemiologia Y Salud Pública (CIBERESP), Barcelona, Spain.
| |
Collapse
|
141
|
Chapman R, van Diepen M, Douglass N, Galant S, Jaffer M, Margolin E, Ximba P, Hermanus T, Moore PL, Williamson AL. Assessment of an LSDV-Vectored Vaccine for Heterologous Prime-Boost Immunizations against HIV. Vaccines (Basel) 2021; 9:1281. [PMID: 34835214 PMCID: PMC8620012 DOI: 10.3390/vaccines9111281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022] Open
Abstract
The modest protective effects of the RV144 HIV-1 vaccine trial have prompted the further exploration of improved poxvirus vector systems that can yield better immune responses and protection. In this study, a recombinant lumpy skin disease virus (LSDV) expressing HIV-1 CAP256.SU gp150 (Env) and a subtype C mosaic Gag was constructed (LSDVGC5) and compared to the equivalent recombinant modified vaccinia Ankara (MVAGC5). In vitro characterization confirmed that cells infected with recombinant LSDV produced Gag virus-like particles containing Env, and that Env expressed on the surface of the cells infected with LSDV was in a native-like conformation. This candidate HIV-1 vaccine (L) was tested in a rabbit model using different heterologous vaccination regimens, in combination with DNA (D) and MVA (M) vectors expressing the equivalent HIV-1 antigens. The four different vaccination regimens (DDMMLL, DDMLML, DDLMLM, and DDLLMM) all elicited high titers of binding and Tier 1A neutralizing antibodies (NAbs), and some regimens induced Tier 1B NAbs. Furthermore, two rabbits in the DDLMLM group developed low levels of autologous Tier 2 NAbs. The humoral immune responses elicited against HIV-1 Env by the recombinant LSDVGC5 were comparable to those induced by MVAGC5.
Collapse
Affiliation(s)
- Ros Chapman
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Science, University of Cape Town, Cape Town 7925, South Africa; (M.v.D.); (N.D.); (S.G.); (E.M.); (P.X.); (A.-L.W.)
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Michiel van Diepen
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Science, University of Cape Town, Cape Town 7925, South Africa; (M.v.D.); (N.D.); (S.G.); (E.M.); (P.X.); (A.-L.W.)
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Nicola Douglass
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Science, University of Cape Town, Cape Town 7925, South Africa; (M.v.D.); (N.D.); (S.G.); (E.M.); (P.X.); (A.-L.W.)
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Shireen Galant
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Science, University of Cape Town, Cape Town 7925, South Africa; (M.v.D.); (N.D.); (S.G.); (E.M.); (P.X.); (A.-L.W.)
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Mohamed Jaffer
- Electron Microscope Unit, University of Cape Town, Rondebosch 7701, South Africa;
| | - Emmanuel Margolin
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Science, University of Cape Town, Cape Town 7925, South Africa; (M.v.D.); (N.D.); (S.G.); (E.M.); (P.X.); (A.-L.W.)
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Phindile Ximba
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Science, University of Cape Town, Cape Town 7925, South Africa; (M.v.D.); (N.D.); (S.G.); (E.M.); (P.X.); (A.-L.W.)
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Tandile Hermanus
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; (T.H.); (P.L.M.)
- Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Penny L. Moore
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; (T.H.); (P.L.M.)
- Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2050, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Congella, Durban 4013, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Science, University of Cape Town, Cape Town 7925, South Africa; (M.v.D.); (N.D.); (S.G.); (E.M.); (P.X.); (A.-L.W.)
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| |
Collapse
|
142
|
Kim J, Vasan S, Kim JH, Ake JA. Current approaches to HIV vaccine development: a narrative review. J Int AIDS Soc 2021; 24 Suppl 7:e25793. [PMID: 34806296 PMCID: PMC8606871 DOI: 10.1002/jia2.25793] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/30/2021] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION The development of an effective vaccine to protect against HIV is a longstanding global health need complicated by challenges inherent to HIV biology and to the execution of vaccine efficacy testing in the context of evolving biomedical prevention interventions. This review describes lessons learnt from previous efficacy trials, highlights unanswered questions, and surveys new approaches in vaccine development addressing these gaps. METHODS We conducted a targeted peer-reviewed literature search of articles and conference abstracts from 1989 through 2021 for HIV vaccine studies and clinical trials. The US National Library of Medicine's Clinical Trials database was accessed to further identify clinical trials involving HIV vaccines. The content of the review was also informed by the authors' own experience and engagement with collaborators in HIV vaccine research. DISCUSSION The HIV vaccine field has successfully developed multiple vaccine platforms through advanced clinical studies; however, the modest efficacy signal of the RV144 Thai trial remains the only demonstration of HIV vaccine protection in humans. Current vaccine strategies include prime-boost strategies to improve elicitation of immune correlates derived from RV144, combination mosaic antigens, novel viral vectors, antigens designed to elicit broadly neutralizing antibody, new nucleic acid platforms and potent adjuvants to enhance immunogenicity across multiple classes of emerging vaccine candidates. CONCLUSIONS HIV vaccine developers have applied lessons learnt from previous successes and failures to innovative vaccine design approaches. These strategies have yielded novel mosaic antigen constructs now in efficacy testing, produced a diverse pipeline of early-stage immunogens and novel adjuvants, and advanced the field towards a globally effective HIV vaccine.
Collapse
Affiliation(s)
- Jiae Kim
- US Military HIV Research ProgramWalter Reed Army Institute of ResearchSilver SpringMarylandUSA
- Henry M. Jackson Foundation for the Advancement of Military MedicineBethesdaMarylandUSA
| | - Sandhya Vasan
- US Military HIV Research ProgramWalter Reed Army Institute of ResearchSilver SpringMarylandUSA
- Henry M. Jackson Foundation for the Advancement of Military MedicineBethesdaMarylandUSA
| | | | - Julie A. Ake
- US Military HIV Research ProgramWalter Reed Army Institute of ResearchSilver SpringMarylandUSA
| |
Collapse
|
143
|
Buchy P, Buisson Y, Cintra O, Dwyer DE, Nissen M, Ortiz de Lejarazu R, Petersen E. COVID-19 pandemic: lessons learned from more than a century of pandemics and current vaccine development for pandemic control. Int J Infect Dis 2021; 112:300-317. [PMID: 34563707 PMCID: PMC8459551 DOI: 10.1016/j.ijid.2021.09.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 01/04/2023] Open
Abstract
Pandemic dynamics and health care responses are markedly different during the COVID-19 pandemic than in earlier outbreaks. Compared with established infectious disease such as influenza, we currently know relatively little about the origin, reservoir, cross-species transmission and evolution of SARS-CoV-2. Health care services, drug availability, laboratory testing, research capacity and global governance are more advanced than during 20th century pandemics, although COVID-19 has highlighted significant gaps. The risk of zoonotic transmission and an associated new pandemic is rising substantially. COVID-19 vaccine development has been done at unprecedented speed, with the usual sequential steps done in parallel. The pandemic has illustrated the feasibility of this approach and the benefits of a globally coordinated response and infrastructure. Some of the COVID-19 vaccines recently developed or currently in development might offer flexibility or sufficiently broad protection to swiftly respond to antigenic drift or emergence of new coronaviruses. Yet many challenges remain, including the large-scale production of sufficient quantity of vaccines, delivery of vaccines to all countries and ensuring vaccination of relevant age groups. This wide vaccine technology approach will be best employed in tandem with active surveillance for emerging variants or new pathogens using antigen mapping, metagenomics and next generation sequencing.
Collapse
Affiliation(s)
| | | | | | - Dominic E Dwyer
- New South Wales Health Pathology - Institute of Clinical Pathology and Medical Research, Westmead Hospital, New South Wales, Australia.
| | - Michael Nissen
- Consultant in Infectious Diseases, University of Queensland, Brisbane, Australia.
| | - Raul Ortiz de Lejarazu
- Scientific Advisor & Emeritus director at Valladolid NIC (National Influenza Centre) Spain, School of Medicine, Avd Ramón y Cajal s/n 47005 Valladolid, Spain.
| | - Eskild Petersen
- European Society for Clinical Microbiology and Infectious Diseases, Basel, Switzerland; Department of Molecular Medicine, The University of Pavia, Pavia, Italy; Department of Clinical, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
144
|
Lanini S, Capone S, Antinori A, Milleri S, Nicastri E, Camerini R, Agrati C, Castilletti C, Mori F, Sacchi A, Matusali G, Gagliardini R, Ammendola V, Cimini E, Grazioli F, Scorzolini L, Napolitano F, Plazzi MM, Soriani M, De Luca A, Battella S, Sommella A, Contino AM, Barra F, Gentile M, Raggioli A, Shi Y, Girardi E, Maeurer M, Capobianchi MR, Vaia F, Piacentini M, Kroemer G, Vitelli A, Colloca S, Folgori A, Ippolito G, Ottou S, Vita S, Vergori A, D'Abramo A, Petrecchia A, Montaldo C, Scalise E, Grassi G, Casetti R, Bordoni V, Notari S, Colavita F, Meschi S, Lapa D, Bordi L, Murachelli S, Tambasco T, Grillo A, Masone E, Marchioni E, Bardhi D, Porzio O, Cocca F, Murachelli S, Turrini I, Malescio F, Ziviani L, Lawlor R, Poli F, Martire F, Zamboni D, Mazzaferri F. GRAd-COV2, a gorilla adenovirus-based candidate vaccine against COVID-19, is safe and immunogenic in younger and older adults. Sci Transl Med 2021; 14:eabj1996. [PMID: 34698501 DOI: 10.1126/scitranslmed.abj1996] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Simone Lanini
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | | | - Andrea Antinori
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | - Stefano Milleri
- Centro Ricerche Cliniche di Verona srl; 37134, Verona, Italy
| | - Emanuele Nicastri
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | | | - Chiara Agrati
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | - Concetta Castilletti
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | | | - Alessandra Sacchi
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | - Giulia Matusali
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | - Roberta Gagliardini
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | | | - Eleonora Cimini
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | | | - Laura Scorzolini
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | | | - Maria M Plazzi
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | | | - Aldo De Luca
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | | | | | | | | | | | | | - Yufang Shi
- First Affiliated Hospital of Soochow University; Suzhou, 215008, Jiangsu, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences; 200061, Shanghai, China
| | - Enrico Girardi
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | - Markus Maeurer
- Division of Immunotherapy, ImmunoSurgery, Champalimaud Foundation; 1400-038, Lisboa, Portugal.,I Medical Clinic, University of Mainz; 55122, Mainz, Germany
| | - Maria R Capobianchi
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy.,Saint Camillus International University of Health Sciences, 00131, Rome, Italy
| | - Francesco Vaia
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata; 00133, Rome, Italy
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006, Paris, France.,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy; 94805, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou; 75015, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, 17164, Stockholm, Sweden
| | | | | | | | - Giuseppe Ippolito
- Istituto Nazionale per Le Malattie Infettive Lazzaro Spallanzani IRCCS; 00149, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Sarkar S, Spencer DA, Barnette P, Pandey S, Sutton WF, Basu M, Burch RE, Cleveland JD, Rosenberg AF, Rangel-Moreno J, Keefer MC, Hessell AJ, Haigwood NL, Kobie JJ. CD4+ T Cells Are Dispensable for Induction of Broad Heterologous HIV Neutralizing Antibodies in Rhesus Macaques. Front Immunol 2021; 12:757811. [PMID: 34745131 PMCID: PMC8564110 DOI: 10.3389/fimmu.2021.757811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/27/2021] [Indexed: 11/24/2022] Open
Abstract
Induction of broadly neutralizing antibodies (bNAbs) is a major goal for HIV vaccine development. HIV envelope glycoprotein (Env)-specific bNAbs isolated from HIV-infected individuals exhibit substantial somatic hypermutation and correlate with T follicular helper (Tfh) responses. Using the VC10014 DNA-protein co-immunization vaccine platform consisting of gp160 plasmids and gp140 trimeric proteins derived from an HIV-1 infected subject that developed bNAbs, we determined the characteristics of the Env-specific humoral response in vaccinated rhesus macaques in the context of CD4+ T cell depletion. Unexpectedly, both CD4+ depleted and non-depleted animals developed comparable Tier 1 and 2 heterologous HIV-1 neutralizing plasma antibody titers. There was no deficit in protection from SHIV challenge, no diminution of titers of HIV Env-specific cross-clade binding antibodies, antibody dependent cellular phagocytosis, or antibody-dependent complement deposition in the CD4+ depleted animals. These collective results suggest that in the presence of diminished CD4+ T cell help, HIV neutralizing antibodies were still generated, which may have implications for developing effective HIV vaccine strategies.
Collapse
Affiliation(s)
- Sanghita Sarkar
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David A. Spencer
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Philip Barnette
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Shilpi Pandey
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - William F. Sutton
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Madhubanti Basu
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Reuben E. Burch
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John D. Cleveland
- School of Public Health, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Alexander F. Rosenberg
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Javier Rangel-Moreno
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, United States
| | - Michael C. Keefer
- Department of Medicine, Division of Infectious Diseases, University of Rochester Medical Center, Rochester, NY, United States
| | - Ann J. Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Nancy L. Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - James J. Kobie
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
146
|
Shivatare SS, Rachel Cheng TJ, Cheng YY, Shivatare VS, Tsai TI, Chuang HY, Wu CY, Wong CH. Immunogenicity Evaluation of N-Glycans Recognized by HIV Broadly Neutralizing Antibodies. ACS Chem Biol 2021; 16:2016-2025. [PMID: 34649433 PMCID: PMC8526942 DOI: 10.1021/acschembio.1c00375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
While the improved treatment of human immunodeficiency virus type 1 (HIV-1) infection is available, the development of an effective and safe prophylactic vaccine against HIV-1 is still an unrealized goal. Encouragingly, the discovery of broadly neutralizing antibodies (bNAbs) from HIV-1 positive patients that are capable of neutralizing a broad spectrum of HIV-1 isolates of various clades has accelerated the progress of vaccine development in the past few years. Some of these bNAbs recognize the N-glycans on the viral surface gp120 glycoprotein. We have been interested in using the glycan epitopes recognized by bNAbs for the development of vaccines to elicit bNAb-like antibodies with broadly neutralizing activities. Toward this goal, we have identified novel hybrid-type structures with subnanomolar avidity toward several bNAbs including PG16, PGT121, PGT128-3C, 2G12, VRC13, VRC-PG05, VRC26.25, VRC26.09, PGDM1400, 35O22, and 10-1074. Here, we report the immunogenicity evaluation of a novel hybrid glycan conjugated to carrier DTCRM197, a nontoxic mutant of the diphtheria toxin, for immunization in mice. Our results indicated that the IgG response was mainly against the chitobiose motif with nonspecific binding to a panel of N-glycans with reducing end GlcNAc-GlcNAc (chitobiose) printed on the glass slides. However, the IgM response was mainly toward the reducing end GlcNAc moiety. We further used the glycoconjugates of Man3GlcNAc2, Man5GlcNAc2, and Man9GlcNAc2 glycans for immunization, and a similar specificity pattern was observed. These findings suggest that the immunogenicity of chitobiose may interfere with the outcome of N-glycan-based vaccines, and modification may be necessary to increase the immunogenicity of the entire N-glycan epitope.
Collapse
Affiliation(s)
- Sachin S. Shivatare
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Ting-Jen Rachel Cheng
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan
| | - Yang-Yu Cheng
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan
| | - Vidya S. Shivatare
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Tsung-I Tsai
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Hong-Yang Chuang
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan
| | - Chung-Yi Wu
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan
| | - Chi-Huey Wong
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Taipei 115, Taiwan
| |
Collapse
|
147
|
Broad and potent bispecific neutralizing antibody gene delivery using adeno-associated viral vectors for passive immunization against HIV-1. J Control Release 2021; 338:633-643. [PMID: 34509584 DOI: 10.1016/j.jconrel.2021.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022]
Abstract
Broadly neutralizing antibodies (bNAbs) possess favorable safety, and passive immunization using these can prevent or control human immunodeficiency virus type 1 (HIV-1) infection. However, bNAbs generally used for monotherapy (IC80 > 5 μg/mL) have limited breadth and potency and neutralize only 70-90% of all HIV-1 strains. To address the need for broader coverage of the HIV-1 epidemic and enhance the ability of bNAbs to target HIV-1, we fused the single-chain variable antibody fragment (scFv) of bNAbs (PG9, PGT123, or NIH45-46) with full-length ibalizumab (iMab) in an scFv-monoclonal antibody tandem format to construct bispecific bNAbs (BibNAbs). Additionally, we described the feasibility of BibNAb gene delivery mediated by recombinant adeno-associated virus 8 (rAAV8) for generating long-term expression with a single injection as opposed to short-term passive immunization requiring continuous injections. Our results showed that the expressed BibNAbs targeting two distinct epitopes exhibited neutralizing activity against 20 HIV-1 pseudoviruses in vitro. After injecting a single rAAV8 vector, the expression and neutralizing activity of the BibNAbs in serum were sustained for 24 weeks. To the best of our knowledge, very few studies have been published on BibNAb gene delivery using rAAV8 vectors against HIV-1. BibNAb gene delivery using rAAV8 vectors may be promising for passive immunization against HIV-1 infection.
Collapse
|
148
|
Yaseen MM, Abuharfeil NM, Darmani H. The impact of MDSCs on the efficacy of preventive and therapeutic HIV vaccines. Cell Immunol 2021; 369:104440. [PMID: 34560382 DOI: 10.1016/j.cellimm.2021.104440] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/07/2021] [Accepted: 09/03/2021] [Indexed: 12/27/2022]
Abstract
In spite of four decades of research on human immunodeficiency virus (HIV), the virus remains a major health problem, affecting tens of millions of people around the world. As such, developing an effective preventive/protective and therapeutic vaccines against HIV are essential to prevent/limit the continuous spread of the virus as well as to control the disease progression and to completely eradicate the virus from HIV infected patients, respectively. There are several factors that have impeded the development of such vaccines, and we need to gain further insight into these factors in order to enhance our knowledge concerning the proper immune activation pathways in the hope of accelerating the development of the highly sought-after vaccine. Recently, new immune cell populations, namely the myeloid-derived suppressor cells (MDSCs), were added to the battle of HIV infection. Indeed, MDSCs seem to play a central role in determining the efficacy of therapeutic and preventive vaccines, especially because vaccines, in general, enhance immune responses, while as a potent immunosuppressor cell population, MDSCs, in turn, subvert and limit the activation of immune responses. Hence, in this work, we sought to address the role of MDSCs in the context of preventive/protective, as well as, therapeutic HIV vaccines.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Homa Darmani
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
149
|
Plummer MM, Pavia CS. COVID-19 Vaccines for HIV-Infected Patients. Viruses 2021; 13:1890. [PMID: 34696319 PMCID: PMC8540182 DOI: 10.3390/v13101890] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/21/2022] Open
Abstract
Nearly 40 years have passed since the initial cases of infection with the human mmunodeficiency virus (HIV) were identified as a new disease entity and the cause of acquired immunodeficiency disease (AIDS). This virus, unlike any other, is capable of causing severe suppression of our adaptive immune defense mechanisms by directly infecting and destroying helper T cells leading to increased susceptibility to a wide variety of microbial pathogens, especially those considered to be intracellular or opportunistic. After T cells are infected, HIV reproduces itself via a somewhat unique mechanism involving various metabolic steps, which includes the use of a reverse transcriptase enzyme that enables the viral RNA to produce copies of its complementary DNA. Subsequent physiologic steps lead to the production of new virus progeny and the eventual death of the invaded T cell. Fortunately, both serologic and molecular tests (such as PCR) can be used to confirm the diagnosis of an HIV infection. In the wake of the current COVID-19 pandemic, it appears that people living with HIV/AIDS are equally or slightly more susceptible to the etiologic agent, SARS-CoV-2, than the general population having intact immune systems, but they may have more serious outcomes. Limited clinical trials have also shown that the currently available COVID-19 vaccines are both safe and effective in affording protection to HIV/AIDS patients. In this review, we further explore the unique dynamic of HIV/AIDS in the context of the worldwide COVID-19 pandemic and the implementation of vaccines as a protective measure against COVID-19, as well as what immune parameters and safeguards should be monitored in this immunocompromised group following vaccination.
Collapse
Affiliation(s)
- Maria M. Plummer
- Department of Clinical Specialties, Division of Pathology, New York Institute of Technology, NYIT College of Osteopathic Medicine, Old Westbury, NY 11568, USA;
| | - Charles S. Pavia
- Department of Biomedical Sciences, New York Institute of Technology, NYIT College of Osteopathic Medicine, Old Westbury, NY 11568, USA
- Division of Infectious Diseases, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
150
|
Sobia P, Archary D. Preventive HIV Vaccines-Leveraging on Lessons from the Past to Pave the Way Forward. Vaccines (Basel) 2021; 9:vaccines9091001. [PMID: 34579238 PMCID: PMC8472969 DOI: 10.3390/vaccines9091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/05/2022] Open
Abstract
Almost four decades on, since the 1980’s, with hundreds of HIV vaccine candidates tested in both non-human primates and humans, and several HIV vaccines trials later, an efficacious HIV vaccine continues to evade us. The enormous worldwide genetic diversity of HIV, combined with HIV’s inherent recombination and high mutation rates, has hampered the development of an effective vaccine. Despite the advent of antiretrovirals as pre-exposure prophylaxis and preventative treatment, which have shown to be effective, HIV infections continue to proliferate, highlighting the great need for a vaccine. Here, we provide a brief history for the HIV vaccine field, with the most recent disappointments and advancements. We also provide an update on current passive immunity trials, testing proof of the concept of the most clinically advanced broadly neutralizing monoclonal antibodies for HIV prevention. Finally, we include mucosal immunity, the importance of vaccine-elicited immune responses and the challenges thereof in the most vulnerable environment–the female genital tract and the rectal surfaces of the gastrointestinal tract for heterosexual and men who have sex with men transmissions, respectively.
Collapse
Affiliation(s)
- Parveen Sobia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: ; Tel.: +27-(0)-31-655-0540
| |
Collapse
|